EP3133083B1 - Multistep final filtration - Google Patents

Multistep final filtration Download PDF

Info

Publication number
EP3133083B1
EP3133083B1 EP16188558.7A EP16188558A EP3133083B1 EP 3133083 B1 EP3133083 B1 EP 3133083B1 EP 16188558 A EP16188558 A EP 16188558A EP 3133083 B1 EP3133083 B1 EP 3133083B1
Authority
EP
European Patent Office
Prior art keywords
filter
pore size
solution
her2 antibody
combination
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP16188558.7A
Other languages
German (de)
French (fr)
Other versions
EP3133083A1 (en
Inventor
Roberto Falkenstein
Klaus Schwendner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
F Hoffmann La Roche AG
Original Assignee
F Hoffmann La Roche AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by F Hoffmann La Roche AG filed Critical F Hoffmann La Roche AG
Priority to PL16188558T priority Critical patent/PL3133083T3/en
Priority to SI201032002T priority patent/SI3133083T1/en
Priority to EP20156152.9A priority patent/EP3715369A1/en
Publication of EP3133083A1 publication Critical patent/EP3133083A1/en
Application granted granted Critical
Publication of EP3133083B1 publication Critical patent/EP3133083B1/en
Priority to HRP20200588TT priority patent/HRP20200588T1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes

Definitions

  • a method for the final filtration of concentrated polypeptide solutions comprising the combination of two immediately consecutive filtration steps with a first filtration step with a pre-filtration with a filter with a pore size of 3.0 ⁇ m and a main-filtration with a filter with a pore size of 0.8 ⁇ m and a second filtration with a pre-filtration with a filter with a pore size of 0.45 ⁇ m and with a main-filtration with a filter with a pore size of 0.22 ⁇ m.
  • Protein solutions with a concentration of more than 100 g/l are prone to difficulties during the final filtration step, e.g. by having only low transmembrane fluxes or blocking of the employed filter by aggregates or particles formed during the formulation or concentration process or due to added excipients resulting in an increased viscosity of the concentrated solution.
  • a two-stage filter constructed using a membrane with a smooth interior underlaid with a thin, flexible porous membrane supported by a rigid screen support with a ridged expander tube is reported in EP 0 204 836 .
  • a combination of at least two membrane filter units of different membrane material and different filter pore size and filter pore geometries is reported in DE 3 818 860 .
  • a filter combination as reported herein of a combination of a first and second filter, whereby the first filter comprises a pre-filter with a pore size of 3.0 ⁇ m and a main-filter with a pore size of 0.8 ⁇ m and the second filter comprises a pre-filter with a pore size of 0.45 ⁇ m and a main-filter with a pore size of 0.22 ⁇ m, for the final filtration of an immunoglobulin solution prior to active pharmaceutical ingredient preparation.
  • An aspect of the invention is a method for producing an anti-HER2 antibody comprising the following steps
  • kits comprising a first filter comprising a pre-filter with a pore size of 3.0 ⁇ m and a main-filter with a pore size of 0.8 ⁇ m and the second filter comprising a pre-filter with a pore size of 0.45 ⁇ m and a main-filter with a pore size of 0.22 ⁇ m.
  • the first filter has an area that is at most twice the area of the second filter. In another embodiment the first and second filter have about the same total filter area.
  • the immunoglobulin solution has a concentration of from 100 g/l to 300 g/l. In still another embodiment the immunoglobulin solution has a volume of from 3 liter to 100 liter. In another embodiment the immunoglobulin solution comprises a sugar and a surfactant and has a concentration of 125 mg/ml or more and the filtrating is with an applied pressure of 0.7 bar or less.
  • the purifying is with a protein A affinity chromatography step and at least one step selected from cation exchange chromatography, anion exchange chromatography, and hydrophobic interaction chromatography.
  • a combination of two filters or filter units each comprising a pre-filter and a main-filter and each with a specifically selected pore size can be used to filter highly concentrated and viscous, as well as formulated immunoglobulin solutions, i.e. comprising a sugar and a surfactant, during the final packaging step.
  • a first filter comprising a pre-filter an a main-filter with a pore size of 3.0 ⁇ m and 0.8 ⁇ m, respectively, and a second filter comprising a pre-filter and a main-filter with a pore size of 0.45 ⁇ m and 0.22 ⁇ m, respectively is highly advantageous.
  • the immunoglobulin solution comprises the immunoglobulin and an excipient.
  • the excipient comprises one or more substances selected from sugars, such as glucose, galactose, maltose, sucrose, trehalose and raffinose, amino acids, such as arginine, lysine, histidine, ornithine, isoleucine, leucine, alanine, glutamic acid, aspartic acid, glycine, and methionine, salts, such as sodium chloride, potassium chloride, sodium citrate, potassium citrate, sodium phosphate, potassium phosphate, and a surfactant, such as polysorbates, and poly (oxyethylene-polyoxypropylene) polymers.
  • the filtrating as reported herein is used as the final filtration step in the production of a therapeutic antibody. It can be carried out after the required excipients, stabilizer and/or anti-oxidants have been added to the highly concentrated antibody solution.
  • the ratio of amount of antibody in kg to total area of the filter is of from 1000 g/m 2 to 10,000 g/m 2 . In another embodiment the ratio is of from 1000 g/m 2 to 6000 g/m 2 . In still another embodiment the ratio is from 4000 g/m 2 to 6000 g/m 2 .
  • polypeptide is a polymer consisting of amino acids joined by peptide bonds, whether produced naturally or synthetically. Polypeptides of less than about 20 amino acid residues may be referred to as "peptides", whereas molecules consisting of two or more polypeptides or comprising one polypeptide of more than 100 amino acid residues may be referred to as "proteins".
  • a polypeptide may also comprise non-amino acid components, such as carbohydrate groups, metal ions, or carboxylic acid esters. The non-amino acid components may be added by the cell, in which the polypeptide is expressed, and may vary with the type of cell. Polypeptides are defined herein in terms of their amino acid backbone structure or the nucleic acid encoding the same. Additions such as carbohydrate groups are generally not specified, but may be present nonetheless.
  • immunoglobulin refers to a protein consisting of one or more polypeptide(s) substantially encoded by immunoglobulin genes.
  • the recognized immunoglobulin genes include the different constant region genes as well as the myriad immunoglobulin variable region genes. Immunoglobulins may exist in a variety of formats, including, for example, Fv, Fab, and F(ab) 2 as well as single chains (scFv) or diabodies.
  • complete immunoglobulin denotes an immunoglobulin which comprises two so called light immunoglobulin chain polypeptides (light chain) and two so called heavy immunoglobulin chain polypeptides (heavy chain).
  • Each of the heavy and light immunoglobulin chain polypeptides of a complete immunoglobulin contains a variable domain (variable region) (generally the amino terminal portion of the polypeptide chain) comprising binding regions that are able to interact with an antigen.
  • variable domain variable region
  • Each of the heavy and light immunoglobulin chain polypeptides of a complete immunoglobulin also comprises a constant region (generally the carboxyl terminal portion).
  • the constant region of the heavy chain mediates the binding of the antibody i) to cells bearing a Fc gamma receptor (Fc ⁇ R), such as phagocytic cells, or ii) to cells bearing the neonatal Fc receptor (FcRn) also known as Brambell receptor. It also mediates the binding to some factors including factors of the classical complement system such as component (C1q).
  • Fc ⁇ R Fc gamma receptor
  • FcRn neonatal Fc receptor
  • C1q component
  • the variable domain of an immunoglobulin's light or heavy chain in turn comprises different segments, i.e. four framework regions (FR) and three hypervariable regions (CDR).
  • immunoglobulin fragment denotes a polypeptide comprising at least one domain of the variable domain of a heavy chain, the C H 1 domain, the hinge-region, the C H 2 domain, the C H 3 domain, the C H 4 domain of a heavy chain, the variable domain of a light chain and/or the C L domain of a light chain. Also comprised are derivatives and variants thereof. For example, a variable domain, in which one or more amino acids or amino acid regions are deleted, may be present.
  • immunoglobulin conjugate denotes a polypeptide comprising at least one domain of an immunoglobulin heavy or light chain conjugated via a peptide bond to a further polypeptide.
  • the further polypeptide is a non-immunoglobulin peptide, such as a hormone, or growth receptor, or antifusogenic peptide, or complement factor, or the like.
  • the term "filter” denotes both a microporous or macroporous filter.
  • the filter comprises a filter membrane which itself is composed of a polymeric material such as, e.g. polyethylene, polypropylene, ethylene vinyl acetate copolymers, polytetrafluoroethylene, polycarbonate, poly vinyl chloride, polyamides (nylon, e.g. ZetaporeTM, N 66 PosidyneTM), polyesters, cellulose acetate, regenerated cellulose, cellulose composites, polysulphones, polyethersulfones, polyarylsulphones, polyphenylsulphones, polyacrylonitrile, polyvinylidene fluoride, non-woven and woven fabrics (e.g.
  • the filter membrane of the first and second filter is made of cellulose acetate.
  • the final purification step is a so called “polishing step" for the removal of trace impurities and contaminants like aggregated immunoglobulins, residual HCP (host cell protein), DNA (host cell nucleic acid), viruses, or endotoxins.
  • polishing step often an anion exchange material in a flow-through mode is used.
  • affinity chromatography with microbial proteins e.g. protein A or protein G affinity chromatography
  • ion exchange chromatography e.g. cation exchange (carboxymethyl resins), anion exchange (amino ethyl resins) and mixed-mode exchange
  • thiophilic adsorption e.g. with beta-mercaptoethanol and other SH ligands
  • hydrophobic interaction or aromatic adsorption chromatography e.g. with phenyl-sepharose, aza-arenophilic resins, or m-aminophenylboronic acid
  • metal chelate affinity chromatography e.g.
  • Ni(II)- and Cu(II)-affinity material size exclusion chromatography
  • electrophoretical methods such as gel electrophoresis, capillary electrophoresis
  • the immunoglobulin solution has a volume of from 3 liter to 100 liter. This solution volume is equivalent to a total mass of the immunoglobulin of from 300 g to 50,000 g. In one embodiment the volume is of from 3.1 liter to 80 liter. At a protein concentration of from 120 g/l to 165 g/l this solution volume is equivalent to a total mass of the immunoglobulin of from 370 g to 13,200 g.
  • An aspect of the invention is a method for producing an anti-HER2 antibody comprising the following steps
  • the cell is a prokaryotic cell or a eukaryotic cell. In one embodiment in which the cell is a prokaryotic cell the cell is selected from E.coli cells, or bacillus cells. In one embodiment in which the cell is a eukaryotic cell the cell is selected from mammalian cells, in a special embodiment from CHO cells, BHK cells, HEK cells, Per.C6® cells and hybridoma cells. In one embodiment the cell is a mammalian cell selected from CHO-K1 and CHO DG44. In one embodiment the cultivating is at a temperature of from 20 °C to 40 °C, and for a period of from 4 to 28 days. In one embodiment the purifying is with a protein A affinity chromatography step and at least one step selected from cation exchange chromatography, anion exchange chromatography, and hydrophobic interaction chromatography.
  • a combination of a first filter unit comprising a pre-filter an a main-filter with a pore size of 3.0 ⁇ m and 0.8 ⁇ m, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 ⁇ m and 0.22 ⁇ m, respectively, is advantageous for processing (filtrating) highly concentrated immunoglobulin solution by allowing the filtration of a complete batch of a concentrated immunoglobulin solution without the need to replace the filter.
  • each of the two filters employed in the units as well as the filter combination has approximately the same filter area, i.e. within two times the area of the smallest filter.
  • the method is operated with an applied pressure of 1.45 bar or more, in another of 1.5 bar or more. If the solution is a concentrated immunoglobulin solution with a concentration of 125 g/l or more, i.e. 130 g/l or 135 g/l, to which at least a sugar and a surfactant have been added then the method is operated in an embodiment with an applied pressure of 0.7 bar or less.
  • kits comprising a first filter unit comprising a pre-filter and a main-filter with a pore size of 3.0 ⁇ m and 0.8 ⁇ m, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 ⁇ m and 0.22 ⁇ m, respectively.
  • a filter comprising a first filter unit comprising a pre-filter and a main-filter with a pore size of 3.0 ⁇ m and 0.8 ⁇ m, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 ⁇ m and 0.22 ⁇ m, respectively for the filtration of a concentrated immunoglobulin solution with a protein concentration of at least 100 g/l.
  • An exemplary antibody is an immunoglobulin against the IL13 receptor ⁇ 1 protein (anti-IL13R ⁇ 1 antibody) e.g. as reported in SEQ ID NO: 01 to 12 of WO 2006/072564 .
  • Another exemplary immunoglobulin is an anti-HER2 antibody reported in WO 92/022653 , WO 99/057134 , WO 97/04801 , US 5,677,171 and US 5,821,337 .
  • Protein A ELISA The wells of a micro titer plate are coated with a polyclonal anti-protein A-IgG derived from chicken. After binding non-reacted antibody is removed by washing with sample buffer. For protein A binding a defined sample volume is added to the wells. The protein A present in the sample is bound by the chicken antibody and retained in the wells of the plate. After the incubation the sample solution is removed and the wells are washed. For detection are added subsequently a chicken derived polyclonal anti-protein A-IgG-biotin conjugate and a Streptavidin peroxidase conjugate.
  • HCP Host cell protein
  • ELISA Host cell protein
  • the walls of the wells of a micro titer plate are coated with a mixture of serum albumin and Streptavidin.
  • a goat derived polyclonal antibody against HCP is bound to the walls of the wells of the micro titer plate.
  • HCP calibration sequence of different concentrations and sample solution.
  • sample material is removed by washing with buffer solution.
  • the wells are incubated with an antibody peroxidase conjugate to detect bound host cell protein.
  • the fixed peroxidase activity is detected by incubation with ABTS and detection at 405 nm.
  • a highly concentrated immunoglobulin solution cannot be filtered with a single sterile filter with a pore size of 0.45 ⁇ m (pre-filter) and 0.22 ⁇ m (main-filter) without blocking of the pores of the filter with a loading of more than 2,460 g protein per square meter of filter area.
  • a highly concentrated immunoglobulin solution can be filtered with a combination of two filters with a pore size of 3.0 ⁇ m (pre-filter) and 0.8 ⁇ m (main-filter) and of 0.45 ⁇ m (pre-filter) and 0.22 ⁇ m (main-filter) without blocking of the pores of the filter independent from the loading of protein per square meter of total filter area.
  • a conditioned protein A eluate can be filtered with a combination of two filters but the flow has to be reduced if the filter area does not match between the two filters.
  • a conditioned protein A eluate can be filtered with a combination of two filters without a reduction of the flow if the filter area does match between the two filters.
  • the filter unit with a pore size of 3.0 ⁇ m and 0.8 ⁇ m has a filter area of 0.2 square meters and the filter unit with a pore size of 0.45 ⁇ m and 0.22 ⁇ m has a filter area of 0.2 square meters.
  • Table 10 Solutions employed in the combined filter filtration. solution No. 16 17 18 19 20 protein mass [g] 495 634 825 861 956 volume [l] 3.5 4.14 5.5 5.6 6.3 loading [g/m 2 ] 1,237.5 1,585.0 2,062.5 2,152.5 2,390
  • Solutions comprising either an anti-IL13R ⁇ antibody or an anti-HER2 antibody were filtered with a filter combination employing different filter area and filter pore size as well as different excipients and applied pressure.
  • Table 13 Results obtained with an anti-HER2 antibody solution with an antibody concentration of 222 mg/ml and an applied pressure of 2.0 bar.

Description

  • Herein is reported a method for the final filtration of concentrated polypeptide solutions comprising the combination of two immediately consecutive filtration steps with a first filtration step with a pre-filtration with a filter with a pore size of 3.0 µm and a main-filtration with a filter with a pore size of 0.8 µm and a second filtration with a pre-filtration with a filter with a pore size of 0.45 µm and with a main-filtration with a filter with a pore size of 0.22 µm.
  • Background of the Invention
  • Protein solutions with a concentration of more than 100 g/l are prone to difficulties during the final filtration step, e.g. by having only low transmembrane fluxes or blocking of the employed filter by aggregates or particles formed during the formulation or concentration process or due to added excipients resulting in an increased viscosity of the concentrated solution.
  • The combination of high viscosity and increased particle or aggregate content results often in the blocking of the pores of an employed 0.22 µm final filtration filter. As a consequence either the filter has to be replaced during the filtration step, i.e. before the batch is completely processed, or an increased filter surface has to be used.
  • Further it has been observed that a combination of a filter with a pore size of 0.45 µm and a filter with a pore size of 0.22 µm has no advantages, e.g. provided as Sartobran P 0.45/0.22 µm filter. Filter with an increased pore size probable to circumvent the before described problems are employed as depth-filters or pre-filters but not a final filters.
  • In DE 4 204 444 a combination of a 1.2 µm pre-filter to remove water droplets from a gas stream prior to a 0.2 µm sterile-filtration is reported. A filter unit comprising two filters of different pore size, whereby the filter of the smaller pore size is flexible allowing by changing the flow direction the filter to bend to reduce the resistance of the filter unit is reported in US 4,488,961 . In US 5,643,566 a combination of a pre-filtration with a filter with a pore size of 0.45 µm and a sterile-filtration with a filter of a pore size of 0.22 µm is reported. A two-stage filter constructed using a membrane with a smooth interior underlaid with a thin, flexible porous membrane supported by a rigid screen support with a ridged expander tube is reported in EP 0 204 836 . A combination of at least two membrane filter units of different membrane material and different filter pore size and filter pore geometries is reported in DE 3 818 860 .
  • Aldington et al. (J. Chrom. B 848 (2007) 64-78) report a scale-up of monoclonal antibody purification processes. In CS 247484 a method of preparing immunoglobulin against human lymphocytes is reported.
  • Summary of the Invention
  • The invention is defined by the claims.
  • It has been found that a combination of two filters each comprising a pre-filter and a main-filter and each with a specifically selected pore size can be used to filter highly concentrated immunoglobulin solutions during the final packaging step without the risk of pore blocking and the need to replace the filter during the filtration process.
  • Disclosed herein is a method for the preparation of an immunoglobulin solution comprising the following steps
    1. a) providing an immunoglobulin solution with a protein concentration of at least 100 g/l,
    2. b) filtering the immunoglobulin solution through a combination of a first and second filter, whereby the first filter comprises a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter comprises a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm, and thereby preparing an immunoglobulin solution.
  • Disclosed herein is the use of a filter combination as reported herein of a combination of a first and second filter, whereby the first filter comprises a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter comprises a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm, for the final filtration of an immunoglobulin solution prior to active pharmaceutical ingredient preparation.
  • An aspect of the invention is a method for producing an anti-HER2 antibody comprising the following steps
    1. a) cultivating a cell comprising a nucleic acid encoding the anti-HER2 antibody,
    2. b) recovering the anti-HER2 antibody from the cell or the cultivation medium,
    3. c) purifying the anti-HER2 antibody with one or more chromatography steps and providing an anti-HER2 antibody solution,
    4. d) adding a sugar and a detergent to the solution,
    5. e) concentrating the anti-HER2 antibody solution to a concentration of 125 g/l or more with a method selected from diafiltration or tangential-flow filtration, and
    6. f) applying the anti-HER2 antibody solution of the previous step to a combination of a first and second filter unit, whereby the first filter unit comprises a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter unit comprises a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm with a pressure of 0.75 bar or less, and thereby producing an anti-HER2 antibody.
  • Disclosed herein is a kit comprising a first filter comprising a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter comprising a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm.
  • In one embodiment the first filter has an area that is at most twice the area of the second filter. In another embodiment the first and second filter have about the same total filter area. In a further embodiment the immunoglobulin solution has a concentration of from 100 g/l to 300 g/l. In still another embodiment the immunoglobulin solution has a volume of from 3 liter to 100 liter. In another embodiment the immunoglobulin solution comprises a sugar and a surfactant and has a concentration of 125 mg/ml or more and the filtrating is with an applied pressure of 0.7 bar or less.
  • In a further embodiment the purifying is with a protein A affinity chromatography step and at least one step selected from cation exchange chromatography, anion exchange chromatography, and hydrophobic interaction chromatography.
  • Detailed Description of the Invention
  • It has been found that a combination of two filters or filter units each comprising a pre-filter and a main-filter and each with a specifically selected pore size can be used to filter highly concentrated and viscous, as well as formulated immunoglobulin solutions, i.e. comprising a sugar and a surfactant, during the final packaging step. Especially the combination of a first filter comprising a pre-filter an a main-filter with a pore size of 3.0 µm and 0.8 µm, respectively, and a second filter comprising a pre-filter and a main-filter with a pore size of 0.45 µm and 0.22 µm, respectively, is highly advantageous. With a single filter unit of this combination it has been possible to filtrate highly concentrated solutions containing in total e.g. 1 kg of an anti-IL-13Ra1 antibody or 6 kg of an anti-HER2 antibody and to package this amounts with only minor substance losses. In one embodiment a ratio of filer surface area to solution volume has been determined.
  • The immunoglobulin solution comprises the immunoglobulin and an excipient. In an embodiment the excipient comprises one or more substances selected from sugars, such as glucose, galactose, maltose, sucrose, trehalose and raffinose, amino acids, such as arginine, lysine, histidine, ornithine, isoleucine, leucine, alanine, glutamic acid, aspartic acid, glycine, and methionine, salts, such as sodium chloride, potassium chloride, sodium citrate, potassium citrate, sodium phosphate, potassium phosphate, and a surfactant, such as polysorbates, and poly (oxyethylene-polyoxypropylene) polymers.
  • The filtrating as reported herein is used as the final filtration step in the production of a therapeutic antibody. It can be carried out after the required excipients, stabilizer and/or anti-oxidants have been added to the highly concentrated antibody solution. In one embodiment the ratio of amount of antibody in kg to total area of the filter is of from 1000 g/m2 to 10,000 g/m2. In another embodiment the ratio is of from 1000 g/m2 to 6000 g/m2. In still another embodiment the ratio is from 4000 g/m2 to 6000 g/m2.
  • A "polypeptide" is a polymer consisting of amino acids joined by peptide bonds, whether produced naturally or synthetically. Polypeptides of less than about 20 amino acid residues may be referred to as "peptides", whereas molecules consisting of two or more polypeptides or comprising one polypeptide of more than 100 amino acid residues may be referred to as "proteins". A polypeptide may also comprise non-amino acid components, such as carbohydrate groups, metal ions, or carboxylic acid esters. The non-amino acid components may be added by the cell, in which the polypeptide is expressed, and may vary with the type of cell. Polypeptides are defined herein in terms of their amino acid backbone structure or the nucleic acid encoding the same. Additions such as carbohydrate groups are generally not specified, but may be present nonetheless.
  • The term "immunoglobulin" refers to a protein consisting of one or more polypeptide(s) substantially encoded by immunoglobulin genes. The recognized immunoglobulin genes include the different constant region genes as well as the myriad immunoglobulin variable region genes. Immunoglobulins may exist in a variety of formats, including, for example, Fv, Fab, and F(ab)2 as well as single chains (scFv) or diabodies.
  • The term "complete immunoglobulin" denotes an immunoglobulin which comprises two so called light immunoglobulin chain polypeptides (light chain) and two so called heavy immunoglobulin chain polypeptides (heavy chain). Each of the heavy and light immunoglobulin chain polypeptides of a complete immunoglobulin contains a variable domain (variable region) (generally the amino terminal portion of the polypeptide chain) comprising binding regions that are able to interact with an antigen. Each of the heavy and light immunoglobulin chain polypeptides of a complete immunoglobulin also comprises a constant region (generally the carboxyl terminal portion). The constant region of the heavy chain mediates the binding of the antibody i) to cells bearing a Fc gamma receptor (FcγR), such as phagocytic cells, or ii) to cells bearing the neonatal Fc receptor (FcRn) also known as Brambell receptor. It also mediates the binding to some factors including factors of the classical complement system such as component (C1q). The variable domain of an immunoglobulin's light or heavy chain in turn comprises different segments, i.e. four framework regions (FR) and three hypervariable regions (CDR).
  • The term "immunoglobulin fragment" denotes a polypeptide comprising at least one domain of the variable domain of a heavy chain, the C H1 domain, the hinge-region, the C H2 domain, the C H3 domain, the C H4 domain of a heavy chain, the variable domain of a light chain and/or the CL domain of a light chain. Also comprised are derivatives and variants thereof. For example, a variable domain, in which one or more amino acids or amino acid regions are deleted, may be present.
  • The term "immunoglobulin conjugate" denotes a polypeptide comprising at least one domain of an immunoglobulin heavy or light chain conjugated via a peptide bond to a further polypeptide. The further polypeptide is a non-immunoglobulin peptide, such as a hormone, or growth receptor, or antifusogenic peptide, or complement factor, or the like.
  • The term "filter" denotes both a microporous or macroporous filter. The filter comprises a filter membrane which itself is composed of a polymeric material such as, e.g. polyethylene, polypropylene, ethylene vinyl acetate copolymers, polytetrafluoroethylene, polycarbonate, poly vinyl chloride, polyamides (nylon, e.g. Zetapore™, N66 Posidyne™), polyesters, cellulose acetate, regenerated cellulose, cellulose composites, polysulphones, polyethersulfones, polyarylsulphones, polyphenylsulphones, polyacrylonitrile, polyvinylidene fluoride, non-woven and woven fabrics (e.g. Tyvek®), fibrous material, or of inorganic material such as zeolithe, SiO2, Al2O3, TiO2, or hydroxyapatite. In one embodiment the filter membrane of the first and second filter is made of cellulose acetate.
  • For the purification of recombinantly produced immunoglobulins often a combination of different column chromatography steps is employed. Generally a protein A affinity chromatography is followed by one or two additional separation steps. The final purification step is a so called "polishing step" for the removal of trace impurities and contaminants like aggregated immunoglobulins, residual HCP (host cell protein), DNA (host cell nucleic acid), viruses, or endotoxins. For this polishing step often an anion exchange material in a flow-through mode is used.
  • Different methods are well established and widespread used for protein recovery and purification, such as affinity chromatography with microbial proteins (e.g. protein A or protein G affinity chromatography), ion exchange chromatography (e.g. cation exchange (carboxymethyl resins), anion exchange (amino ethyl resins) and mixed-mode exchange), thiophilic adsorption (e.g. with beta-mercaptoethanol and other SH ligands), hydrophobic interaction or aromatic adsorption chromatography (e.g. with phenyl-sepharose, aza-arenophilic resins, or m-aminophenylboronic acid), metal chelate affinity chromatography (e.g. with Ni(II)- and Cu(II)-affinity material), size exclusion chromatography, and electrophoretical methods (such as gel electrophoresis, capillary electrophoresis) (Vijayalakshmi, M. A., Appl. Biochem. Biotech. 75 (1998) 93-102).
  • In an embodiment the immunoglobulin solution has a volume of from 3 liter to 100 liter. This solution volume is equivalent to a total mass of the immunoglobulin of from 300 g to 50,000 g. In one embodiment the volume is of from 3.1 liter to 80 liter. At a protein concentration of from 120 g/l to 165 g/l this solution volume is equivalent to a total mass of the immunoglobulin of from 370 g to 13,200 g.
  • An aspect of the invention is a method for producing an anti-HER2 antibody comprising the following steps
    1. a) cultivating a cell comprising a nucleic acid encoding the anti-HER2 antibody,
    2. b) recovering the anti-HER2 antibody from the cell or the cultivation medium,
    3. c) purifying the anti-HER2 antibody with one or more chromatography steps and providing an anti-HER2 antibody solution,
    4. d) adding a sugar and a detergent to the solution,
    5. e) concentrating the anti-HER2 antibody solution to a concentration of 125 g/l or more with a method selected from diafiltration or tangential-flow filtration, and
    6. f) applying the anti-HER2 antibody solution of the previous step to a combination of a first and second filter unit, whereby the first filter unit comprises a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter unit comprises a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm with a pressure of 0.75 bar or less, and thereby producing an anti-HER2 antibody..
  • In one embodiment the cell is a prokaryotic cell or a eukaryotic cell. In one embodiment in which the cell is a prokaryotic cell the cell is selected from E.coli cells, or bacillus cells. In one embodiment in which the cell is a eukaryotic cell the cell is selected from mammalian cells, in a special embodiment from CHO cells, BHK cells, HEK cells, Per.C6® cells and hybridoma cells. In one embodiment the cell is a mammalian cell selected from CHO-K1 and CHO DG44. In one embodiment the cultivating is at a temperature of from 20 °C to 40 °C, and for a period of from 4 to 28 days. In one embodiment the purifying is with a protein A affinity chromatography step and at least one step selected from cation exchange chromatography, anion exchange chromatography, and hydrophobic interaction chromatography.
  • It has been found that a combination of a first filter unit comprising a pre-filter an a main-filter with a pore size of 3.0 µm and 0.8 µm, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 µm and 0.22 µm, respectively, is advantageous for processing (filtrating) highly concentrated immunoglobulin solution by allowing the filtration of a complete batch of a concentrated immunoglobulin solution without the need to replace the filter.
  • It has further been found that in the filter combination it is advantageous that each of the two filters employed in the units as well as the filter combination has approximately the same filter area, i.e. within two times the area of the smallest filter.
  • It has further been found that depending on the components of the solution beside the immunoglobulin different pressure and concentration ranges provide for advantageous processes.
  • If the solution is a concentrated immunoglobulin solution with a concentration of 160 g/l or more, i.e. 165 g/l or 170 g/l, to which no sugar or surfactant has been added then the method is operated with an applied pressure of 1.45 bar or more, in another of 1.5 bar or more. If the solution is a concentrated immunoglobulin solution with a concentration of 125 g/l or more, i.e. 130 g/l or 135 g/l, to which at least a sugar and a surfactant have been added then the method is operated in an embodiment with an applied pressure of 0.7 bar or less.
  • Disclosed herein is a kit comprising a first filter unit comprising a pre-filter and a main-filter with a pore size of 3.0 µm and 0.8 µm, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 µm and 0.22 µm, respectively. Disclosed herein is the use of a filter comprising a first filter unit comprising a pre-filter and a main-filter with a pore size of 3.0 µm and 0.8 µm, respectively, and a second filter unit comprising a pre-filter and a main-filter with a pore size of 0.45 µm and 0.22 µm, respectively for the filtration of a concentrated immunoglobulin solution with a protein concentration of at least 100 g/l.
  • The following examples and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims.
  • Figures
  • Figure 1:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 222 mg/ml and an applied pressure of 2.0 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 2:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with about 200 mM trehalose and about 0.05 % (w/v) Tween 20 and an applied pressure of 2.0 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 3:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 162 mg/ml and an applied pressure of 1.8 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 4:
    Time course of permeate flow obtained with an anti-IL13Rα antibody solution with an antibody concentration of 141 mg/ml supplemented with about 200 mM trehalose and about 0.2 % (w/v) Poloxamer and an applied pressure of 1.6 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 5:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 162 mg/ml and an applied pressure of 1.1 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 6:
    Time course of permeate flow obtained with an anti-IL13Rα antibody solution with an antibody concentration of 141 mg/ml supplemented with trehalose and Poloxamer and an applied pressure of 0.8 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 7:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with trehalose and Tween 20 and an applied pressure of 0.8 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Figure 8:
    Time course of permeate flow obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with trehalose and Tween 20 and an applied pressure of 0.3 bar (diamonds = 1.2 µm pore size filter containing combination; squares = 3.0 µm pore size filter containing combination).
    Example 1 Material and Methods Antibody
  • An exemplary antibody is an immunoglobulin against the IL13 receptor α1 protein (anti-IL13Rα1 antibody) e.g. as reported in SEQ ID NO: 01 to 12 of WO 2006/072564 .
  • Another exemplary immunoglobulin is an anti-HER2 antibody reported in WO 92/022653 , WO 99/057134 , WO 97/04801 , US 5,677,171 and US 5,821,337 .
  • Filter
  • Herein among others a Sartobran P 0.45 µm + 0.2 µm filter cartridge and a Sartoclean CA 3.0 µm + 0.8 µm filter cartridge have been exemplarily employed. Both filter cartridges are available from Sartorius AG, Göttingen, Germany. Analytical Methods
    Size Exclusion Chromatography:
     resin: TSK 3000 (Tosohaas)
     column: 300 x 7.8 mm
     flow rate: 0.5 ml/min
     buffer: 200 mM potassium phosphate containing 250 mM potassium chloride, adjusted to pH 7.0
     wavelength: 280 nm
    DNA-threshold-system: see e.g. Merrick, H., and Hawlitschek, G., Biotech Forum Europe 9 (1992) 398-403
    Protein A ELISA: The wells of a micro titer plate are coated with a polyclonal anti-protein A-IgG derived from chicken. After binding non-reacted antibody is removed by washing with sample buffer. For protein A binding a defined sample volume is added to the wells. The protein A present in the sample is bound by the chicken antibody and retained in the wells of the plate. After the incubation the sample solution is removed and the wells are washed. For detection are added subsequently a chicken derived polyclonal anti-protein A-IgG-biotin conjugate and a Streptavidin peroxidase conjugate. After a further washing step substrate solution is added resulting in the formation of a colored reaction product. The intensity of the color is proportional to the protein A content of the sample. After a defined time the reaction is stopped and the absorbance is measured.
    Host cell protein (HCP) ELISA: The walls of the wells of a micro titer plate are coated with a mixture of serum albumin and Streptavidin. A goat derived polyclonal antibody against HCP is bound to the walls of the wells of the micro titer plate. After a washing step different wells of the micro titer plate are incubated with a HCP calibration sequence of different concentrations and sample solution. After the incubation not bound sample material is removed by washing with buffer solution. For the detection the wells are incubated with an antibody peroxidase conjugate to detect bound host cell protein. The fixed peroxidase activity is detected by incubation with ABTS and detection at 405 nm.
  • Example 2 Filtration of an anti-HER2 antibody with a single filter of 0.45 µm and 0.22 µm pore size
  • In this example it is shown that a highly concentrated immunoglobulin solution cannot be filtered with a single sterile filter with a pore size of 0.45 µm (pre-filter) and 0.22 µm (main-filter) without blocking of the pores of the filter with a loading of more than 2,460 g protein per square meter of filter area.
  • In this example a single filter with a pore size of 0.45 µm and 0.22 µm and a total filter area of 0.2 square meters has been employed. Table 1: Solutions employed in the single filter filtration.
    solution No. 1 2 3 4 5
    protein mass [g] 473 491 496 501 542
    volume [l] 3.940 4.200 4.134 4.139 4.516
    loading [g/m2] 2,365 2,455 2,480 2,505 2,710
  • The concentrated immunoglobulin solutions were filtered through the single filter with the parameters as shown in Table 2. Table 2: Process parameters.
    solution No. 1 2 3 4 5
    volume flow [l/h] 1.97 2.1 Drop to 0 due to pore blocking Drop to 0 due to pore blocking Drop to 0 due to pore blocking
    mass flow [g/h] 237 246 Drop to 0 due to pore blocking Drop to 0 due to pore blocking Drop to 0 due to pore blocking
  • For solutions No. 3 to 5 the pores of the single filter were blocked prior to the complete filtration of the batch volume. To complete the filtration the blocked filter had to be changed resulting in additional time required and loss of product. Table 3: Results of the filtration.
    solution No. 1 2 3 4 5
    protein mass passing the filter [g/m2] 2,365 2,455 960 968 1,440
    volume passing the filter [l] 3.940 4.200 1.600 1.600 2.400
    pore blocking of the filter NO NO YES YES YES
  • Example 3 Filtration of an anti-HER2 antibody with a combination of a first filter with a pore size of 3.0 µm and 0.8 µm and a second filter with a pore size of 0.45 µm and 0.22 µm
  • In this example it is shown that a highly concentrated immunoglobulin solution can be filtered with a combination of two filters with a pore size of 3.0 µm (pre-filter) and 0.8 µm (main-filter) and of 0.45 µm (pre-filter) and 0.22 µm (main-filter) without blocking of the pores of the filter independent from the loading of protein per square meter of total filter area.
  • In this example a combined filter with a first filter unit with a pore size of 3.0 µm and 0.8 µm, respectively, and a second filter unit with a pore size of 0.45 µm and 0.22 µm, respectively, and a filter area each of 0.6 square meters has been employed. Table 4: Solutions employed in the combined filter filtration.
    solution No. 6 7 8 9 10
    protein mass [g] 5,217 5,191 5,356 6,151 5,580
    volume [l] 42.070 42.201 43.542 48.055 44.998
    loading [g/m2] 4,347.5 4,325.8 4,463.3 5,125.8 4,650.0
  • The concentrated immunoglobulin solutions were filtered through the combination of the two filters with the parameters as shown in Table 5. Table 5: Process parameters.
    solution No. 6 7 8 9 10
    volume flow [l/h] 38.95 42.20 43.54 33.02 45.00
    mass flow [g/h] 4830 5191 5356 4226 5580
  • For none of the solutions No. 6 to 10 the pores of the combined filters were blocked prior to the complete filtration of the batch volume. Table 6: Results of the filtration.
    solution No. 6 7 8 9 10
    protein mass passing the filter [g/m2] 4,347.5 4,325.8 4,463.3 5,125.8 4,650.0
    volume passing the filter [l] 42.070 42.201 43.542 48.055 44.998
    pore blocking of the filter NO NO NO NO NO
  • Example 4 Filtration of an anti-IL13Rα antibody with a filter combination of a filter with 3.0 µm and 0.8 µm pore size and a filter with 0.45 µm and 0.22 µm pore size and both filters with different filter areas
  • In this example it is shown that a conditioned protein A eluate can be filtered with a combination of two filters but the flow has to be reduced if the filter area does not match between the two filters.
  • In this example a filter unit with a pore size of 3.0 µm (pre-filter) and 0.8 µm (main-filter) with a filter area of 1.8 square meters and a filter unit with a pore size of 0.45 µm (pre-filter) and 0.22 µm (main-filter) with a filter area of 0.6 square meters has been employed. Table 7: Solutions employed in the combined filter filtration.
    solution No. 11 12 13 14 15
    protein mass [g] 1,169.0 1,299.6 1,154.4 1,220.4 1,284.7
    volume [l] 71.4 76.0 74.0 67.8 70.2
    loading [g/m2] 487.1 541.5 481.0 508.5 535.3
  • The concentrated immunoglobulin solutions were filtered through the combined filter with the parameters as shown in Table 8. Table 8: Process parameters.
    solution No. 11 12 13 14 15
    volume flow [l/h] Drop to 0 due to pore blocking 22 13 12 98
    mass flow [g/h] Drop to 0 due to pore blocking 376 203 216 1793
  • For solution No. 11 the pores of the combined filter were blocked prior to the complete filtration of the batch volume. To complete the filtration the blocked filter had to be changed resulting in additional time required and loss of product. Table 9: Results of the filtration.
    solution No. 1 2 3 4 5
    protein mass passing the filter [g/m2] 347.9 541.5 481.0 508.5 535.3
    volume passing the filter [l] 51.0 76.0 74.0 67.8 70.2
    pore blocking of the filter YES NO NO NO NO
  • In order to prevent filter blocking as in the experiment with solution No. 11 the flow through the membrane had to be reduced in experiments with solutions No. 12 to 14. In experiment with solution No. 15 the protein A eluate has been decanted resulting in a loss of protein.
  • Example 5 Filtration of an anti-IL13Rα antibody with a filter combination of a filter with 3.0 µm and 0.8 µm pore size and a filter with 0.45 µm and 0.22 µm pore size and both filters each with the same filter area
  • In this example it is shown that a conditioned protein A eluate can be filtered with a combination of two filters without a reduction of the flow if the filter area does match between the two filters.
  • In this example the filter unit with a pore size of 3.0 µm and 0.8 µm has a filter area of 0.2 square meters and the filter unit with a pore size of 0.45 µm and 0.22 µm has a filter area of 0.2 square meters. Table 10: Solutions employed in the combined filter filtration.
    solution No. 16 17 18 19 20
    protein mass [g] 495 634 825 861 956
    volume [l] 3.5 4.14 5.5 5.6 6.3
    loading [g/m2] 1,237.5 1,585.0 2,062.5 2,152.5 2,390
  • For none of the solutions No. 16 to 20 the pores of the combined filters were blocked prior to the complete filtration of the batch volume. Table 11: Results of the filtration.
    solution No. 16 17 18 19 20
    Protein mass passing the filter [g/m2] 1,237.5 1,585.0 2,062.5 2,152.5 2,390
    Volume passing the filter [l] 3.5 4.14 5.5 5.6 6.3
    Pore blocking of the filter NO NO NO NO NO
  • Example 6 Filtration of different antibody solutions with different filter combinations with different protein concentrations, different compounds in solution and different applied pressures
  • Solutions comprising either an anti-IL13Rα antibody or an anti-HER2 antibody were filtered with a filter combination employing different filter area and filter pore size as well as different excipients and applied pressure.
  • The used filter combinations are listed in Table 12. In the following the denotation 'A1', 'A2', 'B1', and 'B2' will be used therefore. Table 12: Filter combinations
    combination filter
    1 pore size / diameter filter 2 pore size / diameter filter 3 pore size / diameter filter 4 pore size / diameter
    A1 1.2 µm / 26 mm 0.8 µm / 26 mm 0.45 µm / 26 mm 0.2 µm / 26 mm
    A2 1.2 µm / 47 mm 0.8 µm / 26 mm 0.45 µm / 26 mm 0.2 µm / 26 mm
    B1 3.0 µm / 26 mm 0.8 µm / 26 mm 0.45 µm / 26 mm 0.2 µm / 26 mm
    B2 3.0 µm / 47 mm 0.8 µm / 26 mm 0.45 µm / 26 mm 0.2 µm / 26 mm
  • In the following Tables 13 to 20 and in corresponding Figures 1 to 8 the results obtained with different filter combinations, different antibody solutions and different filtering conditions are presented. Table 13: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 222 mg/ml and an applied pressure of 2.0 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A1 1 3.7 B1 1 3.4
    2 3.5 2 3.2
    3 3.2 3 3.1
    4 3.0 4 3.0
    5 2.9 5 2.8
    6 2.6 6 2.9
    7 2.5 7 2.8
    8 2.3 8 2.7
    9 2.0 9 2.7
    10 2.0 10 2.7
    11 1.7 11 2.7
    12 1.6 12 2.5
    13 1.5 13 2.6
    14 1.3 14 2.5
    15 1.2 15 2.5
    Table 14: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with about 200 mM trehalose and about 0.05 % (w/v) Tween 20 and an applied pressure of 2.0 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A1 1 22.4 B1 1 20.1
    2 20.2 2 17.7
    3 18.3 3 15.5
    4 16.8 4 13.8
    5 15.9 5 12.2
    6 14.3 6 11.1
    7 13.1 7 10.0
    8 12.3 8 8.7
    9 11.3 9 8.1
    10 10.3 10 7.0
    11 9.7 11 6.6
    12 9.2 12 5.8
    13 8.4 13 5.2
    14 8.1 14 4.8
    15 7.4 15 4.3
    Table 15: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 162 mg/ml and an applied pressure of 1.8 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A2 1 7.6 B2 1 8.1
    2 6.5 2 6.9
    3 6.1 3 6.4
    4 5.7 4 6.2
    5 5.4 5 5.9
    6 5.1 6 5.6
    7 5.2 7 5.5
    8 5.0 8 5.3
    9 4.9 9 5.2
    10 4.7 10 5.1
    11 4.8 11 5.0
    12 4.8 12 4.8
    13 4.6 13 4.9
    14 4.7 14 4.6
    15 4.6 15 4.6
    Table 16: Results obtained with an anti-IL13Rα antibody solution with an antibody concentration of 141 mg/ml supplemented with about 200 mM trehalose and about 0.2 % (w/v) Poloxamer and an applied pressure of 1.6 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A2 1 15.6 B2 1 13.2
    2 9.4 2 8.1
    3 7.0 3 5.5
    4 5.5 4 4.1
    5 4.6 5 3.3
    6 3.8 6 2.6
    7 3.3 7 2.3
    8 2.9 8 1.9
    9 2.5 9 1.6
    10 2.2 10 1.5
    11 1.5 11 1.2
    12 0.5 12 1.2
    13 0.3 13 1.0
    14 0.3 14 0.9
    15 0.3 15 0.8
    Table 17: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 162 mg/ml and an applied pressure of 1.1 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A1 1 4.4 B1 1 4.3
    2 4.0 2 4.0
    3 3.6 3 3.5
    4 3.5 4 3.0
    5 3.3 5 3.0
    6 3.2 6 3.0
    7 3.2 7 2.9
    8 3.1 8 2.8
    9 3.1 9 2.8
    10 2.9 10 2.7
    11 3.0 11 2.6
    12 2.9 12 2.8
    13 2.8 13 2.5
    14 2.8 14 2.6
    15 2.8 15 2.5
    Table 18: Results obtained with an anti-IL13Rα antibody solution with an antibody concentration of 141 mg/ml supplemented with trehalose and Poloxamer and an applied pressure of 0.8 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A2 1 7.6 B2 1 8.1
    2 5.0 2 5.5
    3 3.7 3 4.2
    4 2.9 4 3.1
    5 2.5 5 2.6
    6 2.1 6 2.2
    7 1.8 7 1.8
    8 1.5 8 1.5
    9 1.4 9 1.4
    10 1.2 10 1.2
    11 1.1 11 1.1
    12 1.0 12 1.0
    13 0.9 13 0.8
    14 0.8 14 0.8
    15 0.8 15 0.8
    Table 19: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with trehalose and Tween 20 and an applied pressure of 0.8 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A1 1 9.3 B1 1 9.7
    2 8.7 2 8.8
    3 8.1 3 8.4
    4 7.9 4 8.0
    5 7.7 5 7.4
    6 7.2 6 7.0
    7 7.1 7 6.4
    8 6.6 8 6.1
    9 6.2 9 5.7
    10 6.0 10 5.4
    11 5.6 11 5.0
    12 5.3 12 4.6
    13 5.0 13 4.5
    14 4.8 14 4.1
    15 4.5 15 3.3
    Table 20: Results obtained with an anti-HER2 antibody solution with an antibody concentration of 125 mg/ml supplemented with trehalose and Tween 20 and an applied pressure of 0.3 bar.
    combination filtration duration [min] flow [ml/min] combination filtration duration [min] flow [ml/min]
    A1 1 3.9 B1 1 3.7
    2 3.2 2 4.8
    3 3.0 3 4.6
    4 2.7 4 3.8
    5 2.6 5 4.0
    6 2.3 6 3.8
    7 2.1 7 3.8
    8 2.0 8 3.7
    9 1.8 9 3.6
    10 1.5 10 3.6
    11 1.4 11 3.5
    12 1.3 12 3.5
    13 1.2 13 3.3
    14 1.1 14 3.3
    15 1.1 15 3.2

Claims (2)

  1. A method for producing an anti-HER2 antibody comprising the following steps
    a) cultivating a cell comprising a nucleic acid encoding the anti-HER2 antibody,
    b) recovering the anti-HER2 antibody from the cell or the cultivation medium,
    c) purifying the anti-HER2 antibody with one or more chromatography steps and providing an anti-HER2 antibody solution,
    d) adding a sugar and a detergent to the solution,
    e) concentrating the anti-HER2 antibody solution to a concentration of 125 g/l or more with a method selected from diafiltration or tangential-flow filtration, and
    f) applying the anti-HER2 antibody solution of the previous step to a combination of a first and second filter unit, whereby the first filter unit comprises a pre-filter with a pore size of 3.0 µm and a main-filter with a pore size of 0.8 µm and the second filter unit comprises a pre-filter with a pore size of 0.45 µm and a main-filter with a pore size of 0.22 µm with a pressure of 0.75 bar or less, and thereby producing an anti-HER2 antibody.
  2. The method according to claim 1, wherein the purifying is with a protein A affinity chromatography step and at least one step selected from cation exchange chromatography, anion exchange chromatography, and hydrophobic interaction chromatography.
EP16188558.7A 2009-10-01 2010-09-29 Multistep final filtration Active EP3133083B1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
PL16188558T PL3133083T3 (en) 2009-10-01 2010-09-29 Multistep final filtration
SI201032002T SI3133083T1 (en) 2009-10-01 2010-09-29 Multistep final filtration
EP20156152.9A EP3715369A1 (en) 2009-10-01 2010-09-29 Multistep final filtration
HRP20200588TT HRP20200588T1 (en) 2009-10-01 2020-04-14 Multistep final filtration

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP09012460 2009-10-01
EP10757795.9A EP2483305B1 (en) 2009-10-01 2010-09-29 Multistep final filtration of immunoglobulin
PCT/EP2010/064487 WO2011039274A1 (en) 2009-10-01 2010-09-29 Multistep final filtration

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2010/064487 Previously-Filed-Application WO2011039274A1 (en) 2009-10-01 2010-09-29 Multistep final filtration
EP10757795.9A Division EP2483305B1 (en) 2009-10-01 2010-09-29 Multistep final filtration of immunoglobulin

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP20156152.9A Division EP3715369A1 (en) 2009-10-01 2010-09-29 Multistep final filtration

Publications (2)

Publication Number Publication Date
EP3133083A1 EP3133083A1 (en) 2017-02-22
EP3133083B1 true EP3133083B1 (en) 2020-02-19

Family

ID=41466840

Family Applications (3)

Application Number Title Priority Date Filing Date
EP20156152.9A Pending EP3715369A1 (en) 2009-10-01 2010-09-29 Multistep final filtration
EP16188558.7A Active EP3133083B1 (en) 2009-10-01 2010-09-29 Multistep final filtration
EP10757795.9A Active EP2483305B1 (en) 2009-10-01 2010-09-29 Multistep final filtration of immunoglobulin

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP20156152.9A Pending EP3715369A1 (en) 2009-10-01 2010-09-29 Multistep final filtration

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP10757795.9A Active EP2483305B1 (en) 2009-10-01 2010-09-29 Multistep final filtration of immunoglobulin

Country Status (21)

Country Link
US (3) US20120231506A1 (en)
EP (3) EP3715369A1 (en)
JP (1) JP5458183B2 (en)
KR (3) KR101508044B1 (en)
CN (3) CN103980346A (en)
AU (1) AU2010302662B2 (en)
BR (1) BR112012004054B1 (en)
CA (1) CA2773674C (en)
CY (1) CY1118577T1 (en)
DK (1) DK2483305T3 (en)
ES (2) ES2787403T3 (en)
HK (2) HK1200178A1 (en)
HR (2) HRP20161693T1 (en)
HU (1) HUE030186T2 (en)
IL (2) IL217626A (en)
LT (1) LT2483305T (en)
MX (1) MX342442B (en)
PL (2) PL2483305T3 (en)
PT (1) PT2483305T (en)
SI (2) SI3133083T1 (en)
WO (1) WO2011039274A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019183334A1 (en) 2018-03-21 2019-09-26 Waters Technologies Corporation Non-antibody high-affinity-based sample preparation, sorbents, devices and methods
CN111007242A (en) * 2019-12-20 2020-04-14 苏州和迈精密仪器有限公司 Fluorescence immunoassay method and device based on multilayer high molecular porous membrane and application

Family Cites Families (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5643566A (en) 1982-09-23 1997-07-01 Cetus Corporation Formulation processes for lipophilic proteins
US4488961A (en) 1982-09-29 1984-12-18 E. I. Du Pont De Nemours And Company One-way filter unit
US4592848A (en) 1984-12-11 1986-06-03 Pabst Richard E Flow through filter with backflush clearing capability
CS247484B1 (en) 1985-04-12 1987-01-15 Eva Hamsikova Method of immunoglobulin preparation against human lymphocytes
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
DE3818860A1 (en) 1988-06-03 1989-12-07 Seitz Filter Werke FILTER ELEMENT
LU91067I2 (en) 1991-06-14 2004-04-02 Genentech Inc Trastuzumab and its variants and immunochemical derivatives including immotoxins
DE4204444C1 (en) 1992-02-14 1993-05-19 Krankenhausentsorgungs Gmbh, 1000 Berlin, De
PT2275119E (en) 1995-07-27 2013-11-21 Genentech Inc Stable isotonic lyophilized protein formulation
ATE451391T1 (en) 1996-11-27 2009-12-15 Genentech Inc AFFINITY PURIFICATION OF POLYPEPTIDE PROTEINS ON A MATRIX
ES2261589T3 (en) 1998-05-06 2006-11-16 Genentech, Inc. ANTI-HER2 ANTIBODY COMPOSITION.
US6984494B2 (en) * 2000-08-15 2006-01-10 Genentech, Inc. Analytical method
WO2003066662A2 (en) 2002-02-05 2003-08-14 Genentech, Inc. Protein purification
AU2003251592A1 (en) * 2002-06-21 2004-01-06 Biogen Idec Inc. Buffered formulations for concentrating antibodies and methods of use thereof
CN1717483A (en) * 2002-10-04 2006-01-04 麒麟麦酒株式会社 Human artificial chromosome (HAC) vector
WO2004087761A1 (en) 2003-03-31 2004-10-14 Kirin Beer Kabushiki Kaisha Purification of human monoclonal antibody and human polyclonal antibody
GB0316560D0 (en) * 2003-07-15 2003-08-20 Chiron Srl Vesicle filtration
WO2005116072A2 (en) * 2004-05-27 2005-12-08 Weidanz Jon A Antibodies as t cell receptor mimics, methods of production and uses thereof
DK1786273T3 (en) * 2004-08-20 2019-02-18 Prometic Biosciences Ltd SEQUENTIAL PROTEIN INSULATION AND PURIFICATION PLANS WITH AFFINITY CHROMATOGRAPHY
US20060051347A1 (en) * 2004-09-09 2006-03-09 Winter Charles M Process for concentration of antibodies and therapeutic products thereof
AU2005307265A1 (en) * 2004-11-22 2006-05-26 Oxygenix Co., Ltd. Reagent containing oxygen isotope-labeled hemoglobin for examining vital tissue and method of producing the same
TW200902555A (en) 2005-01-03 2009-01-16 Hoffmann La Roche Antibodies against IL-13 receptor alpha 1 and uses thereof
KR100996801B1 (en) * 2005-03-08 2010-11-25 파마시아 앤드 업존 캄파니 엘엘씨 ANTI-MAdCAM ANTIBODY COMPOSITIONS
JPWO2007018316A1 (en) 2005-08-09 2009-02-19 武田薬品工業株式会社 Cancer preventive / therapeutic agent
EP2078039B1 (en) * 2006-11-01 2017-09-13 Biogen MA Inc. Method of isolating biomacromolecules using low ph and divalent cations
WO2009010269A1 (en) * 2007-07-17 2009-01-22 F.Hoffmann-La Roche Ag Variable tangential flow filtration
AR067536A1 (en) 2007-07-17 2009-10-14 Hoffmann La Roche METHOD FOR OBTAINING A MONO-PEGILATED ERYTHROPOYETIN IN A SUBSTANTIALLY HOMOGENOUS FORM
WO2009058769A1 (en) 2007-10-30 2009-05-07 Schering Corporation Purification of antibodies containing hydrophobic variants
CA2739352C (en) 2008-10-29 2021-07-13 Wyeth Llc Methods for purification of single domain antigen binding molecules
JP6345184B2 (en) * 2012-10-30 2018-06-20 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Purification of polypeptides using two-stage tangential flow ultrafiltration

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
IL217626A (en) 2017-12-31
EP3715369A1 (en) 2020-09-30
AU2010302662B2 (en) 2015-11-26
LT2483305T (en) 2016-11-25
PT2483305T (en) 2016-11-04
KR20160104739A (en) 2016-09-05
EP3133083A1 (en) 2017-02-22
US20180009878A1 (en) 2018-01-11
KR20140091763A (en) 2014-07-22
KR20120053522A (en) 2012-05-25
US11891430B2 (en) 2024-02-06
MX2012003727A (en) 2012-04-30
PL3133083T3 (en) 2020-09-07
IL255658A (en) 2018-01-31
HK1210183A1 (en) 2016-04-15
ES2787403T3 (en) 2020-10-16
EP2483305B1 (en) 2016-09-21
US20120231506A1 (en) 2012-09-13
JP5458183B2 (en) 2014-04-02
BR112012004054B1 (en) 2021-09-08
CA2773674A1 (en) 2011-04-07
ES2604103T3 (en) 2017-03-03
SI2483305T1 (en) 2017-01-31
PL2483305T3 (en) 2017-03-31
KR101508044B1 (en) 2015-04-07
BR112012004054A2 (en) 2020-08-11
CY1118577T1 (en) 2017-07-12
US20200291097A1 (en) 2020-09-17
KR101653471B1 (en) 2016-09-01
DK2483305T3 (en) 2016-10-24
CN102574913A (en) 2012-07-11
MX342442B (en) 2016-09-29
HUE030186T2 (en) 2017-04-28
AU2010302662A1 (en) 2012-02-09
HRP20200588T1 (en) 2020-07-10
HK1200178A1 (en) 2015-07-31
CN104610447A (en) 2015-05-13
KR101718301B1 (en) 2017-03-20
EP2483305A1 (en) 2012-08-08
JP2013505733A (en) 2013-02-21
WO2011039274A1 (en) 2011-04-07
HRP20161693T1 (en) 2017-02-24
CA2773674C (en) 2017-08-29
SI3133083T1 (en) 2020-07-31
CN103980346A (en) 2014-08-13

Similar Documents

Publication Publication Date Title
EP2462157B1 (en) Methods for purifying a target protein from one or more impurities in a sample
EP2152745B1 (en) Immunoglobulin purification
CN105646643A (en) Single domain antigen combined molecule purifying method
AU2012282960A1 (en) Method for purifying Fc-fusion protein
US11891430B2 (en) Multistep final filtration
US20220194981A1 (en) Improvement of affinity chromatography of immunoglobulins by using pre-capture flocculation
EP3917494A1 (en) A continuous manufacturing process for biologics manufacturing by integration of drug substance and drug product processes
WO2011038894A1 (en) Protein a chromatography
KR101215740B1 (en) variable tangential flow filtration
US20240132574A1 (en) Multistep final filtration
JP2010051927A (en) Method of separating biogenic substance using ultra-filtration membrane, module and device
CA3203540A1 (en) Systems and methods for process scale isolation of immunoglobulin g
KR20220025873A (en) glycoform tablet
WO2020004583A1 (en) Method for isolating polypeptide, method for producing polypeptide, and apparatus for purifying polypeptide
RU2794431C1 (en) Improving immunoglobulin affinity chromatography by the application of pre-capture floculation
CN113646066A (en) Method for purifying protein

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2483305

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170822

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20180116

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20191009

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 2483305

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602010063203

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1234877

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200315

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: HR

Ref legal event code: TUEP

Ref document number: P20200588

Country of ref document: HR

REG Reference to a national code

Ref country code: RO

Ref legal event code: EPE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20200401047

Country of ref document: GR

Effective date: 20200615

REG Reference to a national code

Ref country code: HR

Ref legal event code: T1PR

Ref document number: P20200588

Country of ref document: HR

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200519

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200619

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200519

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20200588

Country of ref document: HR

Payment date: 20200918

Year of fee payment: 11

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2787403

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20201016

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200712

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602010063203

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20201120

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200929

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20200588

Country of ref document: HR

Payment date: 20210907

Year of fee payment: 12

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 1234877

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200219

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200219

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20200588

Country of ref document: HR

Payment date: 20220909

Year of fee payment: 13

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230414

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20230822

Year of fee payment: 14

REG Reference to a national code

Ref country code: HR

Ref legal event code: ODRP

Ref document number: P20200588

Country of ref document: HR

Payment date: 20230829

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: TR

Payment date: 20230828

Year of fee payment: 14

Ref country code: RO

Payment date: 20230901

Year of fee payment: 14

Ref country code: IT

Payment date: 20230822

Year of fee payment: 14

Ref country code: IE

Payment date: 20230823

Year of fee payment: 14

Ref country code: GB

Payment date: 20230823

Year of fee payment: 14

Ref country code: AT

Payment date: 20230823

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SI

Payment date: 20230908

Year of fee payment: 14

Ref country code: SE

Payment date: 20230822

Year of fee payment: 14

Ref country code: PL

Payment date: 20230824

Year of fee payment: 14

Ref country code: HR

Payment date: 20230829

Year of fee payment: 14

Ref country code: GR

Payment date: 20230823

Year of fee payment: 14

Ref country code: FR

Payment date: 20230822

Year of fee payment: 14

Ref country code: DE

Payment date: 20230822

Year of fee payment: 14

Ref country code: BE

Payment date: 20230822

Year of fee payment: 14

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602010063203

Country of ref document: DE

Representative=s name: KRAUS & LEDERER PARTGMBB, DE

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231002

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: CH

Payment date: 20231001

Year of fee payment: 14